WO2010057015A1 - Structures de rein et leurs procedes de formation - Google Patents

Structures de rein et leurs procedes de formation Download PDF

Info

Publication number
WO2010057015A1
WO2010057015A1 PCT/US2009/064421 US2009064421W WO2010057015A1 WO 2010057015 A1 WO2010057015 A1 WO 2010057015A1 US 2009064421 W US2009064421 W US 2009064421W WO 2010057015 A1 WO2010057015 A1 WO 2010057015A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
anemia
epo
kidney
population
Prior art date
Application number
PCT/US2009/064421
Other languages
English (en)
Inventor
Anthony Atala
James J. Yoo
Original Assignee
Wake Forest University Health Sciences
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wake Forest University Health Sciences filed Critical Wake Forest University Health Sciences
Publication of WO2010057015A1 publication Critical patent/WO2010057015A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0684Cells of the urinary tract or kidneys
    • C12N5/0686Kidney cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/22Urine; Urinary tract, e.g. kidney or bladder; Intraglomerular mesangial cells; Renal mesenchymal cells; Adrenal gland
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/02Atmosphere, e.g. low oxygen conditions

Definitions

  • the present invention is in the field of tissue regeneration for the restoration of organ function.
  • Chronic renal failure is characterized b ⁇ a gradual loss in kidney function, and may eventually progress to end stage renal failure, where the kidney no longer functions at a level to sustain the body.
  • End stage renal failure is a devastating disease that involves multiple organs in affected indh iduals.
  • the most common cause of end stage renal disease in the U.S. is diabetes.
  • EPO erythropoietin
  • the kidney is the primary producer of EPO in the body and is therefore a primary target of treatment for renal failure induced anemia.
  • dialysis can prolong survival for many patients with end stage renal disease, only renal transplantation can currently restore normal function.
  • renal transplantation is severely limited by a critical donor shortage.
  • Treatments used to alleviate anemia associated with renal failure over the >ears include repeated transfusions of red blood cells and administration of testosterone and other anabolic steroids. However, none of these modalities has been entirely satisfactory. Patients receiving repeated transfusions are subject to iron overload, and may develop antibodies to major histocompatibility antigens. Testosterone has a minimal effect on erythropoeisis in the bone marrow, and it is associated with undesirable, virilizing side effects.
  • Renal cell-based approaches to the replacement of kidney tissue is limited by the need to identify and expand renal cells in sufficient quantities.
  • the culturing of renal cells for the purpose of kidney tissue engineering is particularly difficult, owing to the kidney's unique structural and cellular heterogeneity.
  • the kidney is a complex organ with multiple functions, including waste excretion, body homeostasis, electrolyte balance, solute transport, as well as hormone production.
  • isolated populations of cells comprising differentiated mammalian kidney cells, wherein said cells are harvested from mammalian kidney tissue, passaged in vitro, and, optionally, seeded onto a three dimensional matrix.
  • cells are positive for a marker selected from the group consisting of: erythropoietin (EPO). neprilysin (NEP). Tamm-Horsfall protein (THP). podocin (POD), and combinations thereof, after said passaging
  • the population consists essentiall) of said differentiated mammalian kidney cells.
  • the matrix comprises neutralized collagen (e.g.. type I).
  • the mammalian kidney cells have been passaged in vitro from 1 to
  • the mammalian kidney cells have been passaged in vitro at least 3 times. In some embodiments, the mammalian kidney cells have been passaged in vitro from 1 to 5 times.
  • the population has been selected for EPO production (e.g.. prior to having been seeded onto said matrix).
  • Some embodiments are subject to the proviso that the cells are not transfected with an exogenous DNA encoding a polypeptide.
  • composition comprising the population of cells as given above and a pharmaceutically acceptable carrier is also provided.
  • compositions comprising the population of cells as given above:
  • the kidney disease is an anemia selected from the group consisting of: an anemia of renal failure, an anemia of end-stage renal disease, an anemia of a chemotherapy, an anemia of a radiation therapy, an anemia of chronic infection, an anemia of an autoimmune disease, an anemia of rheumatoid arthritis, an anemia of AIDS, an anemia of a malignancy, an anemia of prematurity, an anemia of hypothyroidism, an anemia of malnutrition, and an anemia of a blood disorder.
  • the administering step is carried out by implanting said composition into said patient.
  • Also provided are methods of producing a three dimensional seeded matrix of differentiated kidney cells including providing differentiated kidney cells; and passaging the differentiated kidney cells, wherein the cells are positive for a marker selected from the group consisting of: erythropoietin (EPO), neprilysin (NEP), Tamm-Horsfall protein (THP), podocin (POD), and combinations thereof, after said passaging; seeding said differentiated kidney cells onto a three dimensional matrix; and then solidifying said collagen matrix.
  • the differentiated kidney cells are selected for EPO production.
  • the passaging is carried out from 1 to 20 times.
  • the matrix comprises neutralized collagen (e.g., type I).
  • Methods of treating a kidney disease resulting in decreased EPO production in a patient in need thereof including providing a composition comprising an isolated population of EPO producing cells in a three dimensional matrix; and administering said composition to said patient, whereby said EPO producing cells produce EPO in vivo.
  • the administering step is carried out by implanting said composition into the kidney of said patient.
  • the kidney disease is an anemia selected from the group consisting of: an anemia of renal failure, an anemia of end-stage renal disease, an anemia of a chemotherapy, an anemia of a radiation therapy, an anemia of chronic infection, an anemia of an autoimmune disease, an anemia of rheumatoid arthritis, an anemia of AIDS, an anemia of a malignancy, an anemia of prematurity, an anemia of hypothyroidism, an anemia of malnutrition, and an anemia of a blood disorder.
  • the matrix comprises neutralized collagen (e.g., type I).
  • Another aspect of the present invention is the use of the methods as described herein for the preparation of a composition or medicament for use in treatment or for carrying out a method of treatment as described herein (e.g., for treating a kidney disease or other ailment resulting in decreased EPO production), or for making an article of manufacture as described herein.
  • FIG. 1 Mechanism of erythropoietin (EPO) production. Renal interstitial peritubular cells of the kidney detect low blood oxygen levels, and EPO is secreted into the blood. EPO stimulates the proliferation and differentiation of erythroid progenitors into reticulocytes, and prevents apoptosis, causing more reticulocytes to enter the circulating blood. The reticulocytes differentiate into erythrocytes, increasing the erythron size. Oxygen delivery to the tissues is thereby increased.
  • EPO erythropoietin
  • FIG. 1 Microscopy images of erythropoietin expressing cells in kidney tissue (left panel) and in cultured kidney cells (right panel).
  • FIG. 4 Quantification of erythropoietin (EPO) producing cells. The number of cells expressing EPO decreased with the subsequent passages (* p ⁇ 0.05).
  • Figure 5. Western blot analysis of detergent-solubilized cell extracts detected EPO protein (34 kDa) of early passage primary cultured renal cells (P0-P3).
  • Figure 7A-7B Mouse renal cell characterization. EPO expression is confirmed by immunofluorescence (Figure 7A) (KNRK cells were used as positive control). GLEPPl and Tamm Horsfall kidney markers were also detected ( Figure 7B).
  • FIG 8. Rat renal cell characterization. Cultured rat kidney cells have various cell morphologies shown by phase contrast microscope (left panels), and express GLEPPl and Tamm Horsfall kidney markers (right panels). Figure 9. EPO expression in HepG2 cells was shown by western blot and compared with
  • FIG. 12 Total protein lysates were prepared from rat renal primary cells at passages 1 and 2. Plates from normoxic samples (NC), samples in 3%O2 and 7%O2 were processed and run on 10% SDS-PAGE. KNRK cell line was used as positive control.
  • FIG. 13 Measuring EPO in media concentrates by western blot. Primary cultured cells from Lewis rats were raised close to confluency at each passage on 10cm plates. The cells were starved with KSFM for 24hrs and then placed in a hypoxic chamber (1% 02) for 24, 48 or 72 hrs. Following hypoxia incubation, the media was collected and concentrated with a 1OK mwco amicon ultra centrifugal device (Millipore). 40ug of total protein was then loaded on a 10% polyacrylamide gel. KNRK cells were used as positive control.
  • IxIO 6 cells/injection right panel: Initial cell density of IxIO 5 cells/injection. Top row of each panel: 2 weeks. Bottom row of each panel: 4 weeks.
  • FIG. 15 Effect of culture media and hypoxia on renal primary cells measured by real time PCR. Renal primary cells (p ⁇ ) were grown to 80% confluency in 10cm plates. Three plates of cells were grown with either serum free KSFM or DMEM and placed in a hypoxic chamber at
  • PCR was done in triplicate, and samples were quantified relative to normoxic sample.
  • FIG. 16 Effect of hypoxia on renal primary cells measured by real time PCR. Renal primary cells (passages 0 and 2) were grown to 80% confluency in 10cm plates. Cells were then grown in serum free KSFM and placed in a hypoxic chamber at 1% 02. After 24, 48 or 72 hrs, samples were processed for total RNA and cDNA synthesis. Real time PCR was done in triplicate, and samples were quantified relative to normoxic sample.
  • FIG. 17 Effect of hypoxia on renal primary cells measured by real time PCR. Renal primary cells (passage 0) were grown to 80% confluency in 10cm plates. Cells were then placed in a hypoxic chamber at l%02 for up to 24hrs. Samples were then processed for total RNA and cDNA synthesis. Real time PCR was done in triplicate, and samples were quantified relative to normoxic sample
  • Figure 18 Primary human kidney cells were expanded. Shown are cells of passages 2, 4, 7 and 9. Figure 19. Human primary renal cells were maintained through 20 doublings.
  • Figure 20 Human kidney cell characterization. GLEPPl and EPO positive cells are present in the population.
  • Figure 21 Human kidney cell delivery in vivo with a 20 mg/ml collagen carrier. At retrieval, 3 weeks after injection, the injection volume had been maintained, and neovascularization was present.
  • Figure 22 Injection of collagen with cultured human kidney cells resulted in EPO expressing tissue formation in vivo.
  • Figure 23 Human kidney cell growth curve. Human kidney cells obtained from different donors were cultured and analyzed: hO human kidney cells from 0-year-old (2 months): h65 human kidney cells from a 65-year-old donor: h71 human kidney cells from 71 -year-old donor: and h60 human kidney cells from 60-year-old donor with chronic kidney disease.
  • FIG. 24 Human kidney cell characterization. Left: immunocytochemistry of human kidney cells shows the expression of erythropoietin (A-C). neprilysin (D-F). Tamm-Horsfall protein (G-I). and podocin (J-L) in human kidney cells, passages 1. 2 and 3. Right: western blot analysis of human kidney cells confirmed the expression of the cell-specific proteins at passages
  • Figure 25 Formed renal structures were characterized. A: expression of erythropoietin. neprih sin and Tamm-Horsfall protein were expressed in the newly formed structures. B: western blot analysis confirmed the expression using the same proteins in Panel A. C: E-cadherin was expressed in the formed structures. D: E-cadherein in the structures. E: Na-K ATPase expression.
  • Kidney tissue is tissue isolated or harvested from the kidney, which tissue contains kidney cells. In some embodiments, kidney cells are positive for one or more known kidney markers, e.g.. GLEPPl. Tamm Horsfall. etc.
  • Cell or “cells” may be of any suitable species, and in some embodiments are of the same species as the subject into which tissues produced by the processes herein are implanted. Mammalian cells (including mouse, rat. dog. cat.
  • monkey and human cells are in some embodiments particularly preferred.
  • isolated as used herein signifies that the cells aie placed into conditions other than their natural environment.
  • Tissue or cells are "harvested" when initially isolated from a subject, e.g.. a primary explant. Harvesting of kidney tissue may be performed in accordance with methods known in the art. See also U.S. Patent Application Publication No. 2004/0167634 (Atala et al). which is incorporated by reference herein.
  • Subjects are generally mammalian, including human, subjects and include, but are not limited to. "patients. " The subjects may be male or female and human subjects may be of any race or ethnicity, including, but not limited to. Caucasian. African-American. African. Asian. Hispanic. Indian, etc. The subjects may be of any age. including newborn, neonate, infant, child. adolescent, adult, and geriatric.
  • Subjects and patients may also include animal subjects, particularly mammalian subjects such as canines, felines, bovines. caprines. equines. ovines. porcines. rodents (e.g.. rats and mice), lagomorphs. non-human primates, etc.. for. e.g.. veterinary medicine and/or pharmaceutical drug development purposes.
  • Cells may be syngeneic (i.e.. genetically identical or closely related, so as to minimize tissue transplant rejection), allogeneic (i.e.. from a non-genetically identical member of the same species) or xenogeneic (i.e.. from a member of a different species).
  • Syngeneic cells include those that are autogeneic (i.e...
  • Cells may be obtained from. e.g.. a donor (either living or cadaveric) or derived from an established cell strain or cell line. Cells may be harvested from a donor, e.g.. using standard biopsy techniques known in the art.
  • the “primary culture” is the first culture to become established after seeding disaggregated cells or primary explants into a culture vessel.
  • Expanding refers to an increase in number of viable cells. Expanding may be accomplished by. e.g.. “growing " the cells through one or more cell cycles, wherein at least a portion of the cells divide to produce additional cells.
  • Passaged in vitro refers to the transfer or subculture of a cell culture to a second culture vessel, usually implying mechanical or enzymatic disaggregation, reseeding. and often division into two or more daughter cultures, depending upon the rate of proliferation. If the population is selected for a particular genotype or phenotype. the culture becomes a "cell strain " upon subculture, i.e.. the culture is homogeneous and possesses desirable characteristics (e.g.. the ability to express EPO).
  • EPO expresses or "expression” of EPO means that a gene encoding EPO is transcribed, and preferably, translated. Typically, according to the present invention, expression of an EPO coding region will result in production of the encoded polypeptide, such that the cell is an "EPO producing cell. "
  • cells produce EPO without further manipulation such as the introduction of an exogenous gene.
  • the invention is subject to the proviso that the EPO producing cells are not manipulated by the introduction of an exogenous gene and/or by an exogenous chemical that stimulates the production of EPO.
  • harvested cells are not passaged. In other embodiments, cells are passaged once, twice, or three times. In still other embodiments, cells are passaged more than 3 times.
  • cells are passaged 0-1. 0-2 or 0-3 times. In some embodiments, cells are passaged 1-2. 1-3, or 1-4 or more times. In some embodiments, cells are passaged 2-3 or 2-4 or more times. In further embodiments, cells are passaged 5. 8. 10. 12 or 15 or more times. In some embodiments, cells are passaged 0. 1. 2. 3 or 4 to 8. 10. 15 or 20 or more times.
  • the number of passages used may be selected by. e.g.. the relathe EPO production measured in the cell population after each passage.
  • kidney cells are grown in media that includes additives such as growth factors and other supplements that promote their growth. Further, in some embodiments. EPO producing cells are grown in co-culture with other renal cell types In some embodiments, kidney cells are grown in Dulbecco's Modified Eagle's Medium
  • kidney cells are grown in keratinocyte serum-free medium (KSFM).
  • KSFM keratinocyte serum-free medium
  • kidney cells are grown in KSFM with one or more of the following additives: bovine pituitary extract (BPE) (e.g.. 50g/mL). epidermal growth factor (EGF) (e.g.. 5ng/mL). antibiotic-antimycotic solution (GIBCO) (e.g.. 5 mL).
  • BPE bovine pituitary extract
  • EGF epidermal growth factor
  • GEBCO antibiotic-antimycotic solution
  • FBS fetal bovine serum
  • FBS Gibmini Bio-Product
  • kidney cell are grown in media that is a 1 :1 mixture of keratinocyte serum-free medium (KSFM) and premixed Dulbecco ' s Modified Eagle ' s Medium (DMEM) based media.
  • KSFM keratinocyte serum-free medium
  • DMEM Dulbecco ' s Modified Eagle ' s Medium
  • the premixed DMEM based media is 3 A DMEM and 1 A HAM ' s F12 nutrient mixture supplemented with 5% fetal bovine serum (FBS). 1% Penicillin/Streptomycin. 1% glutamine 10Ox. 1 ml of 0.4 ⁇ g/ml hydrocortisone. 0.5 ml of a 10 "10 M cholera toxin solution. 0.5 ml of a 5 ms/ml insulin solution. 12.5 ml/500ml of a 1.2 mg/ml adenine solution. 0.5 ml of a 2.5 mg/ml transferrin+0.136 mg/ml triiodothyronine mixture, and 0.5ml of a 10 ⁇ g/ml epidermal growth factor (EGF) solution.
  • EGF epidermal growth factor
  • Passaging of kidney cells may be accomplished using standard procedures known in the art.
  • the cells may be detached using trypsin/EDTA and transferred to other plates. This is a standard procedure for many cell types. Briefly, in some embodiments this may be accomplished with the following steps: 1) Remove medium. 2) Add 10 ml PBS/EDTA (0.5 M) for 4 minutes. Confirm the separation of cell junctions under a phase contrast microscope. 3) Remove PBS/EDTA and add 7 ml Trypsin/EDTA. 4) Add 5 ml medium when 80-90% of the cells lift under microscope. 5) Aspirate the cell suspension into a 15 ml test tube. 6) Centrifuge the cells at 1000 rpm for 4 minutes. 7) Remove the supernatant.
  • immunomagnetic bead sorting magnetic activated cell sorting (MASC). panning, etc.
  • MSC magnetic activated cell sorting
  • Unique metabolic pathways and nutritional requirements may be exploited by varying the makeup and/or quantity of nutritional ingredients of the medium on which cells are grown, particularly in a serum-free environment. Protein expression and/or excretion may be detected with various assays, e.g.. ELISA.
  • EPO producing cell refers to differentiated cells, of which at least a portion produce EPO (e.g.. at least 20. 30. 40. or 50% or more, or more preferably 60. 70. 80. or 90% or more of the cells produce EPO).
  • cells produce EPO without further manipulation such as the introduction of an exogenous gene.
  • the invention is subject to the proviso that the EPO producing cells are not manipulated by the introduction of an exogenous gene and/or by an exogenous chemical that stimulates the production of EPO.
  • the cells may be harvested from. e.g.. the peritubular interstitial cells of the kidney. In some embodiments, the cells are selected for their ability to produce EPO.
  • the cells are expanded in number by cell culture techniques, e.g.. passaging.
  • Cells with the specific function of EPO production can be used from the kidney and from other sources.
  • EPO is also normally produced in the liver.
  • EPO is generally known to be produced by the interstitial peritubular cells ( Figure 1).
  • an isolated population of differentiated kidney cells comprises, consists of or consists essentially of interstitial peritubular cells of the kidney, consisting of or consisting essentially of 80. 90. 95. or 99 percent or more, or not more than 20. 10. 5 or 1 percent or less, by number of other cell types.
  • the isolated population of differentiated kidney cells includes other cell types, e.g... endothelial peritubular cells.
  • the isolated population of differentiated kidney cells comprises, consists of or consists essentially of kidney cells that are selected for EPO production, consisting of or consisting essentially of 80. 90. 95. or 99 percent or more, or not more than 20. 10. 5 or 1 percent or less, b ⁇ number of cells not expressing EPO. Selection may be accomplished by selecting the cells that express EPO using specific markers.
  • cells may include various types of kidney cells, so long as the cells express EPO.
  • the entire renal cell colony may be used for expansion and treatment.
  • the isolated population of differentiated kidney cells have a
  • the cells are capable of proliferating through 40. 50 or 60 population doublings or more when grown in vitro.
  • “Differentiated” refers to cells or a population containing cells that have specialized functions, e.g.. EPO production and/or expression of known markers of differentiated cells (e.g.. GLEPPl and/or Tamm Horsfall kidney cell markers). In this sense they are not progenitor or stem cells. Some embodiments of the present invention are subject to the proviso that harvested differentiated cells are not passaged under conditions to create a population of less specialized cells. Alternatively, in other embodiments, cells are cultured to produce cell lines, which may later be differentiated to produce more specialized cells. The establishment of "cell lines. " as opposed to cell strains, are by and large undifferentiated, though they may be committed to a particular lineage.
  • Propagation naturally favors the proliferative phenotype. and in some embodiments cells may require a reinduction of differentiation by. e.g.. alteration of the culture conditions.
  • differentiation factors e.g.. cytokines such as epimorphin and HGF. vitamins, etc.
  • Renal Cell 3D Culture e.g., IL-12, etc.
  • cells are cultured in a three dimensional (3D) matrix.
  • 3D matrix is useful for. inter alia, culturing the cells prior to their administration in methods of treatment. and as an in vitro model system for physiological and developmental studies and/or testing the effects of drugs and/or nephrotoxicity.
  • Three dimensional matrix refers to a matrix of sufficient length, width and height dimensions to allow cells seeded therein to grow and organize therethrough to form a tissue (an aggregate of cells of a particular kind together with their intercellular substance, forming one of the structural materials found in an animal).
  • the matrix material is a polymeric matrix.
  • suitable polymers include, but are not limited to. collagen. poly(alpha esters) such as poly(lactate acid). poly(glycolic acid), polyorthoesters and polyanhydrides and their copolymers, cellulose ether. cellulose, cellulosic ester, fluorinated polyethylene, phenolic, poly-4-methylpentene. polyacrylonitrile. polyamide. polyamideimide. polyacrylate. polybenzoxazole. polycarbonate. pohc ⁇ anoarylether. polyestercarbonate. polyether. polyetheretherketone. polyetherimide. pohetherketone. pol>ethersulfone. polyethylene, polyfluoroolefin.
  • polylmide polyolefin. polyoxadiazole. polyphenylene oxide, polyphenylene. sulfide, polypropylene, polystyrene. polysulfide. polysulfone. polytetrafluoroethylene. polythioether. polytriazole. polyurethane. polyvinylidene fluoride, regenerated cellulose, urea- formaldehyde, or copolymers or physical blends of these materials.
  • the matrix comprises a gel (e.g.. a hydrogel) (optionally solidified after seeding).
  • Hydrogel compositions can include, without limitation, for example, poly(esters). poly(hydroxy acids), poly(lactones). poly(amides). poly(ester-amides). poly(amino acids). poly(anhydrides). poly(ortho-esters). poly(carbonates). poly(phosphazines). poly(thioesters). polysaccharides and mixtures thereof
  • the compositions can also include, for example, a poly(hydroxy) acid including poly(alpha-hydroxy) acids and poly(beta-hydroxy) acids.
  • Such poly(hydroxy) acids include, for example, polylactic acid, polyglycolic acid. polycaproic acid, polybutyric acid, polyvaleric acid, and copolymers and mixtures thereof.
  • the matrix comprises, consists of or consists essentially of collagen. In some embodiments, the matrix comprises, consists of or consists essentially of neutralized Type I collagen. Cells may be seeded onto the matrix and incubated in conditions conducive to their growth in accordance with tissue culture methods described herein and known in the art.
  • the matrix has a length, width and height such that one of these dimensions is not substantially smaller than the other two. as opposed to a thin sheet or flat surface.
  • the thiee dimensional matrix has a length, width and height that do not differ from one another by more than a factor of 2. 3. 4 or 5.
  • the three dimensional culture matrix is created by mixing a collagen type I solution (BD Sciences) and 1OX Medium 199 in a 9:1 ratio, respectively.
  • a base such as NaOH is added until the medium color turns from yellow to red (about 23 ⁇ L IN NaOH for a 1 mL gel solution).
  • cells are seeded onto the matrix by mixing the cells and neutralized collagen solution, e.g.. in a 5:1. 4:1. 3:1. 2:1. 1 :1. 1 :2. 1 :3. 1:4 or 1 :5 ratio. respectively, by volume.
  • the mixing is performed on ice. or at a temperature between 0 and 20 degrees Celsius.
  • the mixture is then gelled (solidified) by incubation at approximately 37 degrees Celsius (e.g.. for 10. 20. 30 or 40 minutes).
  • the mixture may optionally be transferred on to a tissue culture dish prior to gelling.
  • Medium is added to the seeded matrix after solidifying, and changed as needed (e.g.. even 2 days).
  • EPO producing cells are administered to a subject in need thereof
  • EPO producing cells are administered to other areas of the body. e.g. the liver, peritoneum, etc.
  • the EPO producing cells are administered subcutaneously. subcapsular. etc. In further embodiments. EPO producing cells are administered by implantation of a substrate (e.g.. a collagen gel scaffold) containing said EPO producing cells described herein. In still other embodiments. EPO producing cells are administered through ⁇ ascular access (e.g.. systemically or locally).
  • a substrate e.g.. a collagen gel scaffold
  • Anemias include, but are not limited to. those associated with renal failure or end-stage renal disease, anemias caused by chemotherapies or radiation, anemias of chronic disorders, e.g.. chronic infections, autoimmune diseases, rheumatoid arthritis. AIDS. malignancies, anemia of prematurity, anemia of hypothyroidism, anemia of malnutrition (e.g.. iron deficiency), and anemias associated with blood disorders.
  • Treating refers to any type of treatment that imparts a benefit to a patient, e.g.. a patient afflicted with or at risk for developing a disease (e.g.. kidney disease, anemia, etc.). Treating includes actions taken and actions refrained from being taken for the purpose of improving the condition of the patient (e.g.. the relief of one or more symptoms), delay in the onset or progression of the disease, etc.
  • Other endocrine systems may benefit from the therapies disclosed herein, for example. vitamin D producing cell therapy or the angiotensin system. See. e.g.. U.S. Patent Application
  • the cells are mixed with or seeded onto a pharmaceutically acceptable carrier prior to administration.
  • a pharmaceutically acceptable carrier means that the compound or composition is suitable for administration to a subject to achieve the treatments described herein, without unduly deleterious side effects in light of the severity of the disease and necessity of the treatment.
  • Such formulations can be prepared using techniques well known in the art. See. e.g.. U.S. Patent Application 2003/0180289: Remington: The Science and Practice of Pharmacy. Alfonso R. Gennaro. editor. 20th ed. Lippincott Williams & Wilkins: Philadelphia. PA. 2000.
  • the carrier may be a solid or a liquid, or both (e.g.. hydrogels). and can be formulated with the cells as a unit-dose formulation.
  • the cells are provided as a suspension in the carrier to reduce clumping of the cells.
  • cells are seeded onto a biodegradable scaffold or matrix.
  • cells are mixed with a suitable gel for administration.
  • suitable gels that may be used in the present invention include, but are not limited to. agars. collagen, fibrin, hydrogels. etc.
  • other support compounds may also be utilized in the present invention.
  • Extracellular matrix analogs for example, may be combined with support gels to optimize or functionalize the gel.
  • One or more growth factors may also be introduced into the cell suspensions. See. e.g.. U.S. Patent Application Publication No. 2007/0116679 (Atala), which is incorporated by reference herein.
  • Formulations of the invention include those for parenteral administration (e.g.. subcutaneous, intramuscular, intradermal, intravenous, intraarterial, intraperitoneal injection) by injection or implantation.
  • parenteral administration e.g.. subcutaneous, intramuscular, intradermal, intravenous, intraarterial, intraperitoneal injection
  • administration is carried out intravascularly. either by simple injection, or by injection through a catheter positioned in a suitable blood vessel, such as a renal artery.
  • administration is carried out by "infusion. " whereby compositions are introduced into the body through a vein (e.g.. the portal vein).
  • administration is carried out as a graft to an organ or tissue to be augmented as discussed above, e.g.. kidney and/or liver.
  • a “biodegradable scaffold or matrix” is any substance not having toxic or injurious effects on biological function and is capable of being broken down into is elemental components by a host.
  • the scaffold or matrix is porous to allow for cell deposition both on and in the pores of the matrix.
  • Such formulations can be prepared by supplying at least one cell population to a biodegradable scaffold to seed the cell population on and/or into the scaffold. The seeded scaffold or matrix ma) then implanted in the body of a recipient subject.
  • cells are administered by injection of the cells (e.g.. in a suitable carrier) directly into the tissue of a subject.
  • cells may be injected into the kidney (e.g.. the subcapsular space of the kidney). Because the functional effects of EPO production will be systemic, cells may also be administered by injection into other tissues (e.g.. the liver. subcutaneously. etc.).
  • Cells may also be delivered systemically.
  • cells are delivered to tissue outside of the kidney (e.g.. the liver), as the outcome of the functional effects of EPO production will be systemic. See. e.g.. the "Edmonton protocol, " an established delivery method, where cells are infused into a patient's portal vein (Shapiro et al. (2000) N Engl J Med 343:230- 238).
  • the cells administered to the subject may be syngeneic (i.e.. genetically identical or closely related, so as to minimize tissue transplant rejection).
  • allogeneic i.e.. from a non-genetically identical member of the same species
  • xenogeneic i.e.. from a member of a different species
  • Syngeneic cells include those that are autogeneic (i.e.. from the subject to be treated) and isogeneic (i.e.. a genetically identical but different subject, e.g.. from an identical twin).
  • Cells may be obtained from, e.g., a donor (either living or cadaveric) or derived from an established cell strain or cell line.
  • a donor e.g. a potential recipient of a bioscaffold graft
  • standard biopsy techniques known in the art may be employed.
  • cells may be harvested from the subject, expanded/selected in vitro, and reintroduced into the same subject (i.e.. autogeneic).
  • cells are administered in a therapeutically effective amount.
  • the therapeutically effective dosage of cells will vary somewhat from subject to subject, and will depend upon factors such as the age. weight, and condition of the subject and the route of delivery. Such dosages can be determined in accordance with procedures known to those skilled in the art.
  • a dosage of IxIO 3 . IxIO 6 or 5xlO 6 up to IxIO 7 . 1x10 or IxIO 9 cells or more per subject may be given, administered together at a single time or given as several subdivided administrations.
  • a dosage of between 1-100x10 cells per kilogram subject body weight can be given, administered together at a single time or given as several subdivided administration.
  • Cells may be administeied according to some embodiments to achieve a target hematocrit range.
  • the ideal or target hematocrit range may vary from subject to subject, depending upon, e.g.. specific comorbidities.
  • the target hematocrit is from 30-40%.
  • the target hematocrit is from 33-38%.
  • the target hematocrit is from 33-36%.
  • hematocrit may be measured and. if desired or necessary, corrected by. e.g.. further implantation of cells and/or other methods known in the art (e.g.. supplementing with recombinant EPO).
  • an agent for inhibiting transplant rejection of the administered cells such as rapamycin. azathioprine. corticosteroids, cyclosporin and/or FK506. in accordance with known techniques. See. e.g.. R. Calne. U.S. Patent Nos. 5.461.058. 5.403.833 and 5.100.899: see also U.S. Patent Nos. 6.455.518. 6.346.243 and 5.321.043.
  • Some embodiments use a combination of implantation and immunosuppression, which minimizes graft rejection. The implantation may be repeated as needed to create an adequate mass of transplanted tissue.
  • EPO erythropoietin
  • the following examples demonstrate that EPO producing cells are present in renal cells harvested from mouse and rat kidneys.
  • cells isolated and expanded using the methods described below include cells expressing EPO at every culture stage examined.
  • Example 1 Expansion of Renal Cell Primary Cultures. Renal cells from 7-10 day old mice C57BL/6 were culture expanded. Minced kidney (1 kidney of mouse) was placed into a 50cc tube with 15ml of collagenase/dispase (0.2mg/ml). The kidney tissue fragments were incubated in a 37°C shaker for 30 min with collagenase/dispase mix (0.2mg/ml: 15ml). Sterile PBS with Gelatin (20ml). was added (with Gelatin (DIFCO) 2mg/ml) to the digestion solution. The mixture was filtered thorough a 70 micron filter to remove undigested tissue fragments.
  • DIFCO Gelatin
  • KSFM medium (10% FBS, 5ml P/S) is used for stromal cells, and KSFM with BPE, EGF, 5ml antibiotic-antimycotic, 12.5ml FBS (Gemini Bio-Product, 2.5%), Insulin Transferrin Selenium (Roche) (50mg for 5L medium) with BPE and EGF for epithelial components. P/S or antibiotic- antimycotic (GIBCO) may also be added. Each tissue was seeded on to a 25mm plate and medium was added (total 3ml).
  • Cells were maintained by changing the medium the next day. and then every 2 days depending on the cell density. Cells were passaged when they were 80-90% confluent by detachment using trypsin/EDTA and transferred to other plates with the following steps: 1) Remove medium. 2) Add 10 ml PBS/EDTA (0.5 M) for 4 minutes. Confirm the separation of cell junctions under a phase contrast microscope. 3) Remove PBS/EDTA and add 7 ml Trypsin/EDTA. 4) Add 5 ml medium when 80-90% of the cells lift under microscope. 5) Aspirate the cell suspension into a 15 ml test tube. 6) Centrifuge the cells at 1000 rpm for 4 minutes. 7) Remove the supernatant.
  • Kidneys from 10 day old male C57BL/6 mice were collected in Krebs buffer solution (Sigma Aldrich, St. Louis, MO USA) containing 10% antibiotic/antimycotic (Gibco Invitrogen, Carlsbad, CA USA) to avoid risk of contamination.
  • the kidneys were immediately transported to a culture hood where the capsule was removed.
  • the medullary region of the kidney was removed, and only the cortical tissue was used to isolate cells that had been previously identified as EPO producing cells (Maxwell et al., Kidney International, 44: 1149, 1993).
  • kidney tissue was minced and enzymatically digested using Liberase Blendzyme (Roche, Mannheim, Germany,) for 25 minutes at 37 degrees Celsius in a shaking water bath. The supernatant was removed and the cell pellet was passed through a 100 ⁇ m cell strainer to obtain a single cell suspension for culture.
  • Liberase Blendzyme Roche, Mannheim, Germany
  • the cells were plated at a density of 5x10 3 cells/ml in 10 cm tissue culture treated plates filled with culture media.
  • the culture media consisted of a mixture of keratinocyte serum-free medium (KSFM) and premixed Dulbecco's Modified Eagle's Medium (DMEM) at a iatio of 1 1.
  • the piemixed DMEM media contained % DMEM and 1
  • the cells were incubated at 37°C under 5% COT with medium change ever ⁇ 3 days, and the cells were subcultured for expansion at a ratio of 1 :3 when confluent.
  • the cells from early passages (1. 2 and 3) were characterized for EPO expression using lmmunocjtochemistry and western blot analysis with specific antibodies (rabbit polyclonal anti-EPO antibodies, sc-7956. Santa Cruz Technologies. Santa Cruz. California).
  • Renal cells were plated in 8-well chamber slides at a density of 3000 cells per well. The cells were incubated at 37 0 C under 5% CO 2 for 24 h to allow attachment. This was followed by fixation with 4% paraformaldehyde for 10 minutes at room temperature. Permeabihzation of cell membranes was performed by adding 0.1% Triton-X 100 in PBS for 3 minutes at room temperature. Cells were then incubated in goat serum for 30 minutes at room temperature After washing, cells were incubated with the primary antibodies for Ih (1 :50) at room temperature. Cells were washed a second time and biotinylated goat polyclonal anti-rabbit antibodies (polyclonal anti rabbit IgG. Vector Laboratories. Inc..
  • Buiiingame. California (1 :200) were added, followed by incubation at room temperature for 45 minutes. Chromogenic detection of EPO followed a final washing step and was performed using the Vector ABC kit according to the manufacturer ' s instructions (Vector Laboratories. Inc.. Burlingame. California). Slides without the primary antibodies served as internal negative controls, and normal mouse renal tissue served as the positive control.
  • Renal cells in culture showed multiple phenotypes under the microscope. The cells reached confluency within 7 to 10 days of plating. Many of the cells observed in the first 3 passages after isolation from the kidney stained positively for EPO. as compared to the negative controls, which showed no background or nonspecific staining ( Figure 2). which indicated that the observed staining was likely due to the presence of EPO in the cultures. The number of cells that stained positively for EPO remained constant throughout the 3 passages studied, even when phenotypic changes were observed in the culture during the same time period. Immunohistochemical staining of kidney tissue indicated a similar amount of EPO expression as that found in cultured cells (Figure 3). The numbei of cells expressing EPO decreased slightl ⁇ with subsequent passages ( Figure 4). This is most likely due to the increased number of passages and loss of cells/function over time and manipulation. However, the relative percentage appears to remain stable after the first passage EPO expression was also confirmed by western blot, shown in Figure 5.
  • Example 3 Mouse and Rat Renal Cell Characterization. FACS analysis was used to quantify the number of EPO-producing cells in the established renal cell cultures at each passage (1-3 passages). The cells were collected by trypsinization and centrifugation. resuspended in media, and passed through a 70 ⁇ m cell strainer to ensure a single cell suspension. After counting the cells, they were spun down and resuspended in PBS at 5-7.5 xlO 3 cells/ tube to remove FBS from the cells The cells were fixed with 2% formaldehyde for 10 minutes at 4 ° C and permeabilized using 100% methanol for 10 minutes at room temperature.
  • the cells were resuspended in 3% goat serum in PBS followed by incubation with the rabbit anti- EPO primary antibody sc-7956 (Santa Cruz Biotechnology. Santa Cruz. California) for 45 minutes on ice.
  • Cells were washed twice with 3% goat serum in PBS prior to incubation with fluorescein isothiocyanate (FITC)-conjugated goat anti-rabbit secondary antibodies for 1 hour.
  • FITC fluorescein isothiocyanate
  • the cells were then washed thoroughl) with 3% serum in PBS and transferred to the FACS machine (FACS Calibur E6204. Becton-Dickinson. Franklin Lakes. New Jersey).
  • Fluorescent activated cell sorting experiments demonstrated that 44% of passage 1 (Pl) cells were EPO positive. This percentage increased to 82% at passage 2 (P2). and then dropped back to 42% at passage 3 (P3). This may indicate that, during the first few days of culture, proliferation of EPO-producing cells and/or upregulation of EPO gene expression occurs in response to the lower oxygen concentration in the media compared to normal living tissue. These responses could then normalize over the next few days, resulting in numbers of EPO-producing cells that are close to those found in renal tissue (Figure 6. top row).
  • the FACS data demonstrate the maintenance of EPO expression over several passages. It should be noted that there was a surge in the number of cells expressing EPO (82%) in the passage 2 culture, which was confirmed by several repeat experiments. Though not wishing to b ⁇ e bound to any particular theory, one possible explanation for this phenomenon could be that EPO expression is an inherent trait of all renal cells that can be turned on and off as needed. In this case, following the abrupt change in survival conditions between the body and the culture plate, the cells may have been driven to express EPO momentarily until stabilization of the culture occurred. Consistent with this, the EPO surge was quickly reversed and passage 3 analyses showed a lower percentage of EPO producing cells (42%). Mouse cell characterization by immunofluorescence confirmed EPO expression (Figure 7A). The population of cells was positive for the kidney cell markers GLEPPl and Tamm Horsfall ( Figure 7B).
  • Rat cell passages 0, 1 and 2 were also analyzed for EPO production using fluorescence activated cell sorting (FACS) ( Figure 6, bottom row). Cultured rat cells had various cell morphologies and were positive for GLEPPl and Tamm Horsfall kidney cell markers ( Figure 8).
  • FACS fluorescence activated cell sorting
  • EPO Exposure of EPO Producing Cultures to Hypoxic Conditions. While maintenance of phenotypic characteristics is essential during cell expansion stages, a critical component that ensures the success of cell therapy is the ability of EPO producing cells to regulate and maintain normal EPO levels.
  • EPO belongs to the hematopoietic cytokine family, and it controls erythropoiesis in bone marrow, and regulates the proliferation, differentiation and survival of erythroid progenitor cells through EPO receptor (EPOR)-mediated signal transduction. EPO is largely produced in the kidney, and when this organ fails, EPO production falls, leading to anemia. EPO expression in the body depends largely on the oxygen tension in the environment surrounding the cells capable of producing EPO. Factors influencing oxygen levels include lack of oxygen in the ambient air and decreased renal blood flow.
  • EPO expressing cells in culture could respond to changing oxygen levels
  • an experiment was performed in which the cells were serum-starved for 24 hours followed by exposing them to various levels of oxygen in vitro.
  • Lewis rat kidney cells and HepG2 (human hepatocellular liver carcinoma cell line) cells were cultured under normal and hypoxic conditions, and EPO production was assessed and confirmed by western blot of cells.
  • EPO presence in the culture medium was also measured and confirmed by analyzing the supernatants from cultured renal cells under normoxic and hypoxic conditions with the double antibody sandwich enzyme-linked immunosorberbent assay using a Quantikine® IVD® Erythropoietin ELISA kit (R&D Systems®, Minneapolis, Minnesota).
  • HepG2 cells were used as positive controls, as they have been previously reported to produce high levels of EPO in culture (Horiguchi et al.. Blood, 96: 3743). EPO expression by HepG2 was confirmed by western blot (Figure 9). All cells were harvested in lysis buffer containing NP-40. Protein concentration in each sample was measured using a Bio-Rad protein assay. 40 ⁇ g total protein was run out on a 10% acrylamide gel using SDS- PAGE. Proteins were then transferred onto a PVDF membrane (Millipore Corp.).
  • EPO antibody (rabbit polyclonal sc-7956, Santa Cruz Biotechnology) was used at 1 :200 and the secondary antibody (goat anti- rabbit 7074. Cell Signaling Technology, Beverly. Massachusetts) was used at 1 :2000.
  • the media was collected and concentrated down to 500ul using an Amicon Ultra centrifugal filter device (Millipore Corporation. Billerica. Massachusetts). Samples of this media were run on a 10% polyacrylamide gel.
  • EPO antibody (rabbit polyclonal sc-7956. Santa Cruz Biotechnology) was used at 1: 100 and the secondary antibody (goat anti-rabbit 7074, Cell Signaling Technology, Beverly, MA, USA) was used at 1 :2000.
  • total protein lysates were prepared from rat renal primary cells at passages 1 and 2. Plates from normoxic samples (NC). samples in 3%O2 and 7%O2 were processed and Run on 10% SDS-PAGE. The KNRK cell line was used as positive control. Results are shown in Figure 12.
  • Example 5 Administration of EPO Producing Cells in vivo.
  • renal cells mixed in collagen gel were implanted subcutaneously in athymic mice at concentrations of 1 xlO 6 and 5 xlO followed by retrieval at 14 and 28 days after implantation for analysis. Cells at different passages from 1-5 were used. The cells were suspended in a collagen gel for easy injection (concentration: O. lmg/ml).
  • EPO producing renal cells grown and expanded in culture stably expressed EPO in vivo.
  • EPO producing cells may be used as a treatment option for anemia caused by chronic renal failure.
  • Example 6 Analysis of EPO Expression with Real Time PCR. Real time PCR was performed to assess rat cell expression of EPO in response to hypoxic conditions.
  • KSFM and DMEM were exposed to hypoxic conditions (3% O 2 ). Renal primary cells (passage O) were grown to 80% confluency in 10cm plates. Three plates of cells were grown with either serum free KSFM or DMEM and placed in a hypoxic chamber at 3% O 2 . After 24 hrs. samples were processed for total RNA and cDNA synthesis. Real time PCR was done in triplicate, and samples were quantified relative to normoxic sample. Results are shown in Figure 15.
  • Rat kidney culture EPO expression was compared with real time PCR across 24, 48 and 72 hours. Renal primary cells (passages 0 and 2) were grown to 80% confluency in 10cm plates. Cells were then grown in serum free KSFM and placed in a hypoxic chamber at 1% 02. After 24. 48 or 72 hours, samples were processed for total RNA and cDNA synthesis. Real time PCR was done in triplicate, and samples were quantified relative to normoxic sample. Results are shown in Figure 16.
  • renal primary cells (passage 0) were grown to 80% confluency in 10cm plates. Cells were then placed in a hypoxic chamber at 1% 02 for up to 24 hours. Samples were then processed for total RNA and cDNA synthesis. Real time PCR w ; as run in triplicate, and samples were quantified relative to normoxic sample. Results are shown in
  • Example 7 Expansion of Human Kidney Cells. The growth and expandability of primary human kidney cells were also demonstrated using the media and conditions described above. Cultures from passages 2, 4, 7 and 9 are shown in Figure 18. It was demonstrated that human primary renal cells can be maintained through twenty doublings (Figure 19). Human kidney cell cultures were characterized for EPO and GLEPPl expression ( Figure 20).
  • Example 8 Human Kidney Cell Delivery Via Collagen Injection. Human renal cells mixed in collagen gel were implanted subcutaneously in athymic mice as described above in Example 5. Collagen concentrations of lmg/ml, 2mg/ml and 20mg/ml were compared. At 1 and 2 mg/ml, the in vivo volume disappeared after administration. At 20 mg/ml, the in vivo injection volume was maintained, and neo-vascularization was seen Figure 21. Histology confirmed that EPO expressing tissue was formed in vivo ( Figure 22).
  • Example 9 EPO Producing Cell Selection with Magnetic Cell Sorting.
  • Cells are selected for EPO production using magnetic cell sorting.
  • a single-cell suspension is isolated using a standard preparation method. After preparation of single-cell suspension, count the total number of the cells and centrifuge cell samples to obtain a pellet. Block the cells with 10% of goat serum (of animal where the secondary antibody is made) for 10 minutes. Add 1 or 2 mL of the blocking solution. After 10 minutes of centrifugation. resuspend the cells in the primary antibody for EPO (use l ⁇ g of the primary antibody/ million of cells). Typically, label for 15 minutes at 4-8 0 C is sufficient.
  • EXAMPLE 10 In Vitro Reconstitution of Human Kidney Structures for Renal Failure. End stage renal disease is currently being treated effectively by transplantation. However, increasing demand and donor shortage make this treatment challenging. Recent advances in cell-based therapies have provided potential opportunities to alleviate the current challenges of donor shortage. We previously have demonstrated that single renal cells expanded in culture are able to form renal structures when implanted in vivo. However, the levels of structure formation could not be adequately controlled. In this study, we investigated whether human kidney structures could be pre-formed in vitro for subsequent implantation in vivo to maximize tissue forming efficiency. Primary human renal cells were isolated from unused donor kidneys using enzymatic digestion methods. Renal cells were grown, expanded and characterized using cell specific antibodies.
  • kidney structures single renal cells were placed in a three-dimensional culture system, consisting of neutralized type I collagen.
  • the three-dimensional matrix with cells was solidified and cultivated over a period of 10 days. Histomorphological and ultrastructural analyses were performed using cell specific markers that identify proximal and distal tubules and collecting ducts.
  • EXAMPLE 11 //; Vivo Implantation of Reconstituted of Human Kidney Structures.
  • Primary human renal cells were isolated from unused donor kidneys using enzymatic digestion methods. Renal cells were grown, expanded and characterized by immunocytochemistry and western blot using cell specific antibodies: proximal tubules: neprilysin (NEP); collecting duct: Tamm-Horsfall protein (THP); podocytes: podocin (POD). The ability of these cells to migrate was analyzed using different growth factors.
  • NEP neprilysin
  • THP Tamm-Horsfall protein
  • podocytes podocin
  • the ability of these cells to migrate was analyzed using different growth factors.
  • To form kidney structures single renal cells were placed in a three-dimensional culture system of neutralized type I collagen. Histomorphological and ultrastructural analyses were performed using cell specific markers that identify proximal and distal tubules and collecting ducts. Albumin uptake assay was used to analyze the functionality of these tubul
  • Renal cells placed in a three-dimensional culture environment began to proliferate and form structures that resemble renal tubules.
  • Immunocytochemistry showed that the reconstituted renal structures were positive for expression of erythropoietin (EPO), Neprilysin (NEP) (proximal tubules), Tamm-Horsfall protein (THP) (collecting duct), and Podocin (POD) markers in passages 1, 2 and 3.
  • EPO erythropoietin
  • NEP Neprilysin
  • THP Tamm-Horsfall protein
  • POD Podocin
  • Western blot analysis confirmed the presence of these protein markers.
  • E- cadherin, N-cadherin and Na-K ATPase staining confirmed polarization of the cells present in the tubules.
  • Co-localization of labeled albumin and tubule markers (including epithelia membrane antigen (EMA). which is a distal tubule marker) proved functionality and specific
  • 3D cultures were implanted in the kidney of nude rats to evaluate survival of the cells (GFP cells) over a period of 6 weeks.
  • the formed structures stained positively for anti-GFP after 3 weeks of implantation.
  • Human kidney cells showed evidence of migration towards native tissue after 6 weeks of implantation in the interstitium area and in glomeruli.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Developmental Biology & Embryology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

L'invention concerne des populations isolées de cellules de rein prélevées à partir de cellules différentiées du rein, lesdites cellules ayant été développées in vitro, ainsi que leurs procédés d'utilisation. Les cellules peuvent être fournies dans une matrice tridimensionnelle en vue de les cultiver in vitro et/ou de les implanter in vivo. L'invention concerne enfin des procédés d'ensemencement de cellules sur ladite matrice.
PCT/US2009/064421 2008-11-14 2009-11-13 Structures de rein et leurs procedes de formation WO2010057015A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19934408P 2008-11-14 2008-11-14
US61/199,344 2008-11-14

Publications (1)

Publication Number Publication Date
WO2010057015A1 true WO2010057015A1 (fr) 2010-05-20

Family

ID=42170365

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/064421 WO2010057015A1 (fr) 2008-11-14 2009-11-13 Structures de rein et leurs procedes de formation

Country Status (2)

Country Link
US (1) US9580688B2 (fr)
WO (1) WO2010057015A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8318484B2 (en) 2008-11-12 2012-11-27 Tengion, Inc. Isolated renal cells and uses thereof
RU2620947C2 (ru) * 2011-05-27 2017-05-30 ДЕПУИ СИНТЕЗ ПРОДАКТС, ЭлЭлСи Биосинтетические системы проксимального канальца и способы использования
US9724367B2 (en) 2010-11-10 2017-08-08 Regenmed (Cayman) Ltd. Injectable formulations for organ augmentation
WO2018013947A1 (fr) 2016-07-14 2018-01-18 Qidni Labs, Inc. Matériau et filtre biocompatibles et hémocompatibles
US10077442B2 (en) 2010-05-12 2018-09-18 Inregen Bioactive renal cells
US11123372B2 (en) 2016-07-29 2021-09-21 Prokidney Bioactive renal cells for the treatment of chronic kidney disease

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2005509495A (ja) * 2001-11-16 2005-04-14 チルドレンズ メディカル センター コーポレーション 臓器の機能の増強
EP2626410A1 (fr) * 2012-02-08 2013-08-14 Technische Hochshule Mittelhessen Dispositif et procédé d'expansion et de récolte de lignées cellulaires adhérentes
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070059293A1 (en) * 2002-02-05 2007-03-15 Children's Medical Center Corporation Tissue engineered construct for supplementing or replacing a damaged organ
WO2008045498A1 (fr) * 2006-10-12 2008-04-17 Ethicon, Inc. Populations de cellules extraites de reins et leur utilisation pour la réparation et le régénération de tissus

Family Cites Families (97)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3463158A (en) 1963-10-31 1969-08-26 American Cyanamid Co Polyglycolic acid prosthetic devices
JPS5492580A (en) 1977-12-29 1979-07-21 Nippon Zeon Co Ltd Hollow fiber type material transferring apparatus
US4182339A (en) 1978-05-17 1980-01-08 Hardy Thomas G Jr Anastomotic device and method
JPS6045849B2 (ja) 1980-08-25 1985-10-12 林原 健 ヒトエリトロポエチンの製造方法
SU1034717A1 (ru) 1981-04-22 1983-08-15 Рижский Медицинский Институт Способ наложени сосудистого анастомоза
US4458678A (en) 1981-10-26 1984-07-10 Massachusetts Institute Of Technology Cell-seeding procedures involving fibrous lattices
US4520821A (en) 1982-04-30 1985-06-04 The Regents Of The University Of California Growing of long-term biological tissue correction structures in vivo
US4594407A (en) 1983-09-20 1986-06-10 Allied Corporation Prosthetic devices derived from krebs-cycle dicarboxylic acids and diols
FR2577807B1 (fr) 1985-02-22 1993-12-03 Ethnor Materiau chirurgical composite absorbable, procede de preparation, prothese resorbable realisee a partir d'un tel materiau et utilisation d'une telle prothese
US5032508A (en) 1988-09-08 1991-07-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5266480A (en) 1986-04-18 1993-11-30 Advanced Tissue Sciences, Inc. Three-dimensional skin culture system
US4963489A (en) 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
US5863531A (en) 1986-04-18 1999-01-26 Advanced Tissue Sciences, Inc. In vitro preparation of tubular tissue structures by stromal cell culture on a three-dimensional framework
US5160490A (en) 1986-04-18 1992-11-03 Marrow-Tech Incorporated Three-dimensional cell and tissue culture apparatus
US5902741A (en) 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US4769037A (en) 1986-10-28 1988-09-06 Midcalf Robert J Artificial replacement kidney implant and method of dialyzing blood
CA1340581C (fr) 1986-11-20 1999-06-08 Joseph P. Vacanti Neomorphogenese chimerique d'organes par implatation cellulaire controlee, utilisant des matrices artificielles
US5759830A (en) 1986-11-20 1998-06-02 Massachusetts Institute Of Technology Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
US5567612A (en) 1986-11-20 1996-10-22 Massachusetts Institute Of Technology Genitourinary cell-matrix structure for implantation into a human and a method of making
WO1989001967A1 (fr) 1987-09-03 1989-03-09 Brown University Research Foundation Purification du sang a l'aide de cellules renales cultivees
GB8722854D0 (en) 1987-09-29 1987-11-04 Hardy S M Implantable artificial kidney
US5085629A (en) 1988-10-06 1992-02-04 Medical Engineering Corporation Biodegradable stent
CN1053698C (zh) 1989-01-19 2000-06-21 南京大学 细胞培养生产促红细胞生成素的方法
DE3913756A1 (de) 1989-04-21 1990-10-25 Schering Ag 8(beta)-substituierte ergoline, verfahren zu ihrer herstellung und ihre verwendung
US5429936A (en) * 1989-04-21 1995-07-04 The Regents Of The University Of California Antibody-mediated juxtaposition of reactive moieties
DE69017820T2 (de) 1989-04-25 1995-10-05 Childrens Medical Center Implantaten für grosse mengen von zellen auf polymerische matrizen.
US4996154A (en) 1989-05-04 1991-02-26 Millipore Corporation Method for growing cellular tissue
WO1991009625A1 (fr) 1989-12-21 1991-07-11 Tanox Biosystems, Inc. Anticorps monoclonaux qui neutralisent l'infection par hiv-1 et leurs anti-idiotypes
US5261898A (en) 1990-11-13 1993-11-16 Polin Stanton G Temporary colostomy apparatus
US5192312A (en) 1991-03-05 1993-03-09 Colorado State University Research Foundation Treated tissue for implantation and methods of treatment and use
WO1993007913A1 (fr) 1991-10-24 1993-04-29 Children's Medical Center Corporation Neomorphogenese in vivo de structures urologiques a partir de cultures de cellules
NZ245015A (en) 1991-11-05 1995-12-21 Transkaryotic Therapies Inc Delivery of human growth hormone through the administration of transfected cell lines encoding human growth hormone, which are physically protected from host immune response; the transfected cells and their production
US5376376A (en) 1992-01-13 1994-12-27 Li; Shu-Tung Resorbable vascular wound dressings
US6410320B1 (en) * 1992-03-02 2002-06-25 The University Of Michigan Method and compositions for isolation and growth of kidney tubule stem cells, in vitro kidney tubulogenesis and ex vivo construction of renal tubules
US5549674A (en) 1992-03-02 1996-08-27 The Regents Of The University Of Michigan Methods and compositions of a bioartificial kidney suitable for use in vivo or ex vivo
US5429938A (en) 1992-03-02 1995-07-04 University Of Michigan Methods and compositions for isolation and growth of kidney tubule stem cells, in vitro kidney tubulogenesis and ex vivo construction of renal tubules
US6060270A (en) 1992-03-02 2000-05-09 The University Of Michigan Methods and compositions for isolation and growth of kidney tubule stem cells, in vitro kidney tubulogenesis and ex vivo construction of renal tubules
US5441870A (en) 1992-04-15 1995-08-15 Athena Neurosciences, Inc. Methods for monitoring cellular processing of β-amyloid precursor protein
CA2118520C (fr) 1992-04-24 1999-08-10 Dirkjan Bakker Dispositifs pour prevenir l'adherence tissulaire
US5224953A (en) 1992-05-01 1993-07-06 The Beth Israel Hospital Association Method for treatment of obstructive portions of urinary passageways
US5800537A (en) 1992-08-07 1998-09-01 Tissue Engineering, Inc. Method and construct for producing graft tissue from an extracellular matrix
US5957972A (en) 1992-09-29 1999-09-28 Arizona Board Of Regents On Behalf Of The University Of Arizona Implants possessing a surface of endothelial cells genetically-modified to inhibit intimal thickening
US5514378A (en) 1993-02-01 1996-05-07 Massachusetts Institute Of Technology Biocompatible polymer membranes and methods of preparation of three dimensional membrane structures
US5433996A (en) 1993-02-18 1995-07-18 W. L. Gore & Associates, Inc. Laminated patch tissue repair sheet material
US5709854A (en) 1993-04-30 1998-01-20 Massachusetts Institute Of Technology Tissue formation by injecting a cell-polymeric solution that gels in vivo
KR100365573B1 (ko) 1994-03-14 2004-08-25 크라이어라이프, 인크. 이식용처리조직및이의제조방법
US5916265A (en) 1994-03-30 1999-06-29 Hu; Jie Method of producing a biological extracellular matrix for use as a cell seeding scaffold and implant
US5626861A (en) 1994-04-01 1997-05-06 Massachusetts Institute Of Technology Polymeric-hydroxyapatite bone composite
US5947893A (en) 1994-04-27 1999-09-07 Board Of Regents, The University Of Texas System Method of making a porous prothesis with biodegradable coatings
US5545131A (en) 1994-04-28 1996-08-13 White Eagle International Technologies, Lp Artificial kidney
US5429674A (en) 1994-09-12 1995-07-04 Ppg Industries, Inc. N-acyl aminomethylene phosphonates and their use in waterborne coating compositions
JPH10511563A (ja) 1994-09-12 1998-11-10 アドバンスド ティシュー サイエンシズ,インコーポレーテッド 心臓弁枠組み上での三次元ヒト細胞培養物およびその使用
US5654273A (en) 1994-09-22 1997-08-05 Children's Medical Center Corporation Synducin mediated modulation of tissue repair
US5695996A (en) 1994-09-23 1997-12-09 The United States Of America As Represented By The Department Of Health And Human Services Artificial organ culture system
US5716404A (en) 1994-12-16 1998-02-10 Massachusetts Institute Of Technology Breast tissue engineering
CA2214407C (fr) 1995-04-07 2008-06-17 Purdue Research Foundation Greffe de tissus et procede de reconstitution de vessie
US5855610A (en) 1995-05-19 1999-01-05 Children's Medical Center Corporation Engineering of strong, pliable tissues
WO1998006445A1 (fr) 1996-08-16 1998-02-19 Children's Medical Center Corporation Sous-muqueuse vesicale ensemencee par des cellules pour la reconstitution de son tissu
US6171344B1 (en) 1996-08-16 2001-01-09 Children's Medical Center Corporation Bladder submucosa seeded with cells for tissue reconstruction
US6673339B1 (en) 1996-09-05 2004-01-06 Children's Medical Center Corporation Prosthetic kidney and its use for treating kidney disease
EP0929271B1 (fr) 1996-09-05 2004-02-25 Children's Medical Center Corporation Rein prothetique
US5952226A (en) 1996-11-05 1999-09-14 Modex Therapeutiques Hypoxia responsive EPO producing cells
US5967972A (en) 1997-03-28 1999-10-19 Kapp Surgical Instrument, Inc. Minimally invasive surgical retractor and method of operation
WO1998046165A1 (fr) 1997-04-11 1998-10-22 Cryolife, Inc. Decellularisation d'un tissu
DE59803925D1 (de) 1997-06-27 2002-05-29 Augustinus Bader Bioartifizielles transplantat und verfahren zu seiner herstellung
ATE328617T1 (de) 1997-10-31 2006-06-15 Childrens Medical Center Penis-rekonstruktion
US6576019B1 (en) 1997-10-31 2003-06-10 Children's Medical Center Corporation Bladder reconstruction
EP0984062A1 (fr) 1998-09-04 2000-03-08 Cytos Biotechnology AG Production d'érythropoeitine humaine
US6777205B1 (en) 1998-11-06 2004-08-17 Sterrenbeld Biotechnologie North America, Inc. Host cells expressing recombinant human erythropoietin
DE19919625C2 (de) 1999-04-29 2002-10-31 Symetis Ag Zuerich In-vitro-Verfahren zum Herstellen einer homologen Herzklappe und durch dieses Verfahren herstellbare Klappe
DE60017900T2 (de) 1999-04-30 2006-04-06 Massachusetts General Hospital, Boston Herstellung von dreidimensionalem vaskularisierten gewebe mittels der verwendung von zweidimensionalen mikrohergestellten formen
GB9910377D0 (en) 1999-05-05 1999-06-30 Univ Westminster Tissue engineering
MXPA01011429A (es) 1999-05-11 2003-09-10 Ortho Mcneil Pharm Inc Modelado farmacocinetico y farmacodinamico de administracion de eritropoyetina.
US20040167634A1 (en) 1999-05-26 2004-08-26 Anthony Atala Prosthetic kidney and its use for treating kidney disease
KR100369788B1 (ko) 1999-09-03 2003-01-29 동아제약 주식회사 재조합 인간 에리트로포이에틴의 제조방법
JP3603179B2 (ja) 1999-09-09 2004-12-22 グンゼ株式会社 心血管系組織培養用基材および組織再生法
US20030180289A1 (en) 1999-09-23 2003-09-25 Foster Keith Alan Inhibition of secretion from non-neuronal cells
US6503330B1 (en) 1999-12-22 2003-01-07 Genus, Inc. Apparatus and method to achieve continuous interface and ultrathin film during atomic layer deposition
US6428802B1 (en) 1999-12-29 2002-08-06 Children's Medical Center Corp. Preparing artificial organs by forming polylayers of different cell populations on a substrate
US6376244B1 (en) 1999-12-29 2002-04-23 Children's Medical Center Corporation Methods and compositions for organ decellularization
US6479064B1 (en) 1999-12-29 2002-11-12 Children's Medical Center Corporation Culturing different cell populations on a decellularized natural biostructure for organ reconstruction
US6368859B1 (en) 1999-12-29 2002-04-09 Children's Medical Center Corporation Methods and compositions for producing a fascial sling
US7326570B2 (en) 2000-06-16 2008-02-05 The Regents Of The University Of California Induction of tubular morphogenesis using pleiotrophin
RU2003109746A (ru) 2000-09-08 2005-01-27 Грифон Терапьютикс, Инк. (Us) Синтетические белки, стимулирующие эритропоэз
WO2002061053A1 (fr) 2001-01-31 2002-08-08 The General Hospital Corporation Cellules souches renales et leurs utilisations
US6656488B2 (en) 2001-04-11 2003-12-02 Ethicon Endo-Surgery, Inc. Bioabsorbable bag containing bioabsorbable materials of different bioabsorption rates for tissue engineering
FR2824072B1 (fr) 2001-04-27 2004-03-12 Centre Nat Rech Scient Procede de replication et de production du virus de l'hepatite c
US6784154B2 (en) 2001-11-01 2004-08-31 University Of Utah Research Foundation Method of use of erythropoietin to treat ischemic acute renal failure
JP2005509495A (ja) 2001-11-16 2005-04-14 チルドレンズ メディカル センター コーポレーション 臓器の機能の増強
EP1543128A4 (fr) 2002-07-18 2008-02-20 Invitrogen Corp Vecteurs viraux contenant des sites de recombinaison
US8940292B2 (en) 2003-01-28 2015-01-27 Wake Forest University Health Sciences Enhancement of angiogenesis to grafts using cells engineered to produce growth factors
WO2004091495A2 (fr) 2003-04-09 2004-10-28 University Of Utah Research Foundation Compositions et procedes associes a la production d'erythropoietine
US20050002919A1 (en) 2003-06-09 2005-01-06 The Brigham And Women's Hospital, Inc. Cadherin directed molecular and cellular localization
AU2003903896A0 (en) 2003-07-28 2003-08-07 Queensland University Of Technology Skin regeneration system
US20050136042A1 (en) 2003-08-12 2005-06-23 Betz Oliver B. Methods and compositions for tissue repair
EP1548031A1 (fr) 2003-12-22 2005-06-29 Dubai Genetics FZ-LLC Erythropoietin identique à la nature
JP2009508650A (ja) 2005-09-21 2009-03-05 ダスク テクノロジーズ, エルエルシー 臓器および組織機能のための方法および組成

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070059293A1 (en) * 2002-02-05 2007-03-15 Children's Medical Center Corporation Tissue engineered construct for supplementing or replacing a damaged organ
WO2008045498A1 (fr) * 2006-10-12 2008-04-17 Ethicon, Inc. Populations de cellules extraites de reins et leur utilisation pour la réparation et le régénération de tissus

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8318484B2 (en) 2008-11-12 2012-11-27 Tengion, Inc. Isolated renal cells and uses thereof
US9192629B2 (en) 2008-11-12 2015-11-24 Regenmedtx, Llc Isolated renal cells and uses thereof
US10105392B2 (en) 2008-11-12 2018-10-23 Inregen Isolated renal cells and uses thereof
US10077442B2 (en) 2010-05-12 2018-09-18 Inregen Bioactive renal cells
US11066666B2 (en) 2010-05-12 2021-07-20 Inregen Bioactive renal cells
US9724367B2 (en) 2010-11-10 2017-08-08 Regenmed (Cayman) Ltd. Injectable formulations for organ augmentation
RU2620947C2 (ru) * 2011-05-27 2017-05-30 ДЕПУИ СИНТЕЗ ПРОДАКТС, ЭлЭлСи Биосинтетические системы проксимального канальца и способы использования
WO2018013947A1 (fr) 2016-07-14 2018-01-18 Qidni Labs, Inc. Matériau et filtre biocompatibles et hémocompatibles
US11123372B2 (en) 2016-07-29 2021-09-21 Prokidney Bioactive renal cells for the treatment of chronic kidney disease
US11931383B2 (en) 2016-07-29 2024-03-19 Prokidney Bioactive renal cells for the treatment of chronic kidney disease

Also Published As

Publication number Publication date
US20100112062A1 (en) 2010-05-06
US9580688B2 (en) 2017-02-28

Similar Documents

Publication Publication Date Title
US9534203B2 (en) Selective cell therapy for the treatment of renal failure
US9580688B2 (en) Kidney structures and methods of forming the same
US10590391B2 (en) Selective cell therapy for the treatment of renal failure
JP7062605B2 (ja) 単離腎細胞を含有するオルガノイド及びその使用
KR100823221B1 (ko) 인간 췌장 상피 기원 세포 및 그것의 분리 및 이용 방법
ES2672205T3 (es) Células madre hepáticas aisladas
CA2779144C (fr) Cellules souches pluripotentes a partir de l'arbre biliaire extrahepatique et procedes pour leur isolement
WO2010065239A1 (fr) Cellules souches provenant d’urine et leurs procédés d’utilisation
ES2359874T3 (es) Células madre hepáticas aisladas.
KR20140042824A (ko) 인공 근위세뇨관 시스템 및 사용 방법
US20220213433A1 (en) Three-Dimensional Cross-Linked Scaffolds of Cord Blood Plasma and Their Use
AU2014240277B2 (en) Selective cell therapy for the treatment of renal failure
EP4023231A1 (fr) Vésicules extracellulaires dérivées de cellules stromales mésenchymateuses génétiquement modifiées pour la surexpression hif-1a et htert
BRPI0620049A2 (pt) células-tronco isoladas do fìgado
WO2023180122A1 (fr) Utilisation de cellules progénitrices allogéniques humaines dérivées du foie pour traiter et/ou prévenir la sénescence cellulaire
NZ713875B2 (en) Organoids comprising isolated renal cells and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09826848

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09826848

Country of ref document: EP

Kind code of ref document: A1